EP2893939A1 - Anti-netrin-1 antibody - Google Patents

Anti-netrin-1 antibody Download PDF

Info

Publication number
EP2893939A1
EP2893939A1 EP14305034.2A EP14305034A EP2893939A1 EP 2893939 A1 EP2893939 A1 EP 2893939A1 EP 14305034 A EP14305034 A EP 14305034A EP 2893939 A1 EP2893939 A1 EP 2893939A1
Authority
EP
European Patent Office
Prior art keywords
netrin
seq
polypeptide
antibody
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP14305034.2A
Other languages
German (de)
English (en)
French (fr)
Inventor
Jean-Guy Delcros
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Netris Pharma SAS
Original Assignee
Netris Pharma SAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Netris Pharma SAS filed Critical Netris Pharma SAS
Priority to EP14305034.2A priority Critical patent/EP2893939A1/en
Priority to MX2016009029A priority patent/MX2016009029A/es
Priority to SG11201605562VA priority patent/SG11201605562VA/en
Priority to PCT/EP2015/050306 priority patent/WO2015104360A1/en
Priority to JP2016545904A priority patent/JP6586095B2/ja
Priority to LTEP15700118.1T priority patent/LT3092003T/lt
Priority to PT157001181T priority patent/PT3092003T/pt
Priority to TW104100790A priority patent/TWI699211B/zh
Priority to DK15700118.1T priority patent/DK3092003T3/da
Priority to PL15700118T priority patent/PL3092003T3/pl
Priority to US15/110,612 priority patent/US10494427B2/en
Priority to SI201531074T priority patent/SI3092003T1/sl
Priority to CN201580007753.8A priority patent/CN105979966B/zh
Priority to KR1020167018645A priority patent/KR102360967B1/ko
Priority to EA201691212A priority patent/EA034676B1/ru
Priority to NZ721975A priority patent/NZ721975A/en
Priority to ARP150100062A priority patent/AR099068A1/es
Priority to HUE15700118A priority patent/HUE048088T2/hu
Priority to ES15700118T priority patent/ES2770620T3/es
Priority to RS20200058A priority patent/RS59818B1/sr
Priority to EP15700118.1A priority patent/EP3092003B1/en
Priority to CA2936308A priority patent/CA2936308C/en
Priority to AU2015205575A priority patent/AU2015205575B2/en
Priority to BR112016015811-3A priority patent/BR112016015811B1/pt
Publication of EP2893939A1 publication Critical patent/EP2893939A1/en
Priority to IL246603A priority patent/IL246603B/en
Priority to PH12016501339A priority patent/PH12016501339A1/en
Priority to ZA2016/04640A priority patent/ZA201604640B/en
Priority to CL2016001753A priority patent/CL2016001753A1/es
Priority to US16/657,200 priority patent/US11648309B2/en
Priority to HRP20192313TT priority patent/HRP20192313T1/hr
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention relates to a netrin-1 binding polypeptide which is useful in inducing cell death or apoptosis of tumor cells bearing a netrin-1 receptor such as an UNC5 receptor in the presence of netrin-1, in particular the tumor cells express (autocrine expression) netrin-1, and its use for the treatment of cancer.
  • a netrin-1 binding polypeptide which is useful in inducing cell death or apoptosis of tumor cells bearing a netrin-1 receptor such as an UNC5 receptor in the presence of netrin-1, in particular the tumor cells express (autocrine expression) netrin-1, and its use for the treatment of cancer.
  • Netrin-1 is a member of the netrin family and displays an axon navigation cue, both, in an attractive and repulsive context and plays a major role in the development of the nervous system.
  • the main receptors for netrin-1 are DCC (Deleted in Colorectal Cancer) and UNC5 (UNC5A, UNC5B, UNC5C, UNC5D in human, UNC5H1, UNC5H2, UNC5H3, UNC5H4 in mice), which all belong to the dependence receptor family ( Keino-Masu, 1996, Cell 87: 175-185 ; Ackermann, 1997, Nature 386: 838-842 ; Hong, 1999, Cell 97: 927-941 ; Mehlen, 1998, Nature 395: 801-804 ).
  • Dependence receptors share the ability to induce apoptosis in the absence of their respective ligands, whereby this ability is blocked upon binding of the respective ligand ( Mehlen, 2004, Cell Mol Life Sci 61: 1854-1866 ; Bredesen, 2005, Cell Death Differ 12: 1031-1043 ).
  • neutralization of netrin-1 by the ectodomain of DCC or part of this ectodomain can induce apoptosis in tumor cells expressing dependence receptors DCC and/or UNC5 ( EP-A1-1 989 546 ).
  • This ectodomain or part of this ectodomain is capable to reduce metastasis of breast cancer cells into the lung (Fitamant et al. 2008).
  • this ectodomain or part of this ectodomain has been also demonstrated to increase the cell death percentage of non-small cell lung cancer cells and neuroblastoma cells expressing high levels of netrin-1 ( Delloye-Bourgeois, 2009, J Natl Cancer Inst 101 : 237-247 ; Delloye-Bourgeois, 2009, JEM 206 : 833-847 ).
  • WO2012025618 discloses DCC-fusion proteins having improved DCC decoy.
  • EP 1 989 546 discloses monoclonal or polyclonal antibodies directed specifically against netrin-1 or netrin-1 receptor, particularly directed to the extracellular domain of the netrin-1 receptor or to the netrin-1 fragment able to interact with the extracellular domain of said netrin-1 receptor, as a medicament.
  • the Applicant has now determined a linear or substantially linear epitope on netrin-1 that likely corresponds to the specific binding region of netrin-1 to receptor, in particular of the UNC5 class, especially UNC5B and UNC5A, or alternatively corresponds to a region nearby the specific binding region of netrin-1 to receptor that when bound to an antibody prevents netrin-1/receptor interaction.
  • This determination of a linear epitope allows the Applicant to produce antibodies binding to netrin-1 and interfering with netrin-1/receptor interaction, thereby inducing apoptosis or cell death of tumour cells expressing or overexpressing netrin-1 and at least one netrin-1 receptor, owing the fact that this interaction inhibits netrin-1 binding to a receptor and the multimerization of the receptor.
  • the Applicant also produced a murine monoclonal antibody directed against this epitope and various humanized forms thereof.
  • the full-length amino acid sequence of netrin-1 is given as SEQ ID NO: 1 and a cDNA coding therefore is given at SEQ ID NO: 2.
  • a linear epitope has been characterized in the second EGF-like domain of netrin-1 and is depicted on SEQ ID NO: 3.
  • a cDNA encoding this epitope is depicted on SEQ ID NO: 4.
  • a first object of the invention is thus a polypeptide representing a linear epitope of netrin-1 or a fragment or variant of said linear epitope. More specifically, the invention relates to:
  • Another object of the invention is a cDNA coding for said polypeptide representing a linear epitope of netrin-1 or a fragment or variant of said linear epitope. More specifically, the invention relates to:
  • a variant polypeptide is able to generate an antibody which still keeps the ability to specifically bind to the linear epitope on netrin-1 and inhibit the interaction of netrin-1 to its receptor, especially UNC5B and UNC5A, and induce apoptosis or cell death of the tumour cell expressing or overexpressing netrin-1 and a netrin-1 receptor.
  • a variant cDNA codes for the polypeptide of SEQ ID NO: 3 or for a variant polypeptide.
  • Another object of the invention is the use of the polypeptide of SEQ ID NO: 3 or a variant thereof, to produce a monoclonal antibody, and the monoclonal antibodies so produced are also an object of the invention.
  • the person skilled in the art is aware of the methods allowing producing monoclonal antibodies, using the plasmocyte and hybridoma technique.
  • the invention encompasses the use of these methods to produce antibodies specifically directed against the polypeptide of SEQ ID NO: 3 or a variant thereof. It is also within the scope of the invention to produce polypeptides or monoclonal antibodies through genetic engineering based on nucleic acid sequences coding for the specific polypeptide or antibody, owing the determination and disclosure of the CDRs of the VH and VL.
  • binding polypeptide will be used herein to encompass antibodies and antibody variants, fragments and combination that keep the binding function of the antibody.
  • the present invention thus also relates to a netrin-1 binding polypeptide which specifically binds to a polypeptide having the amino acid sequence SEQ ID NO: 3 or a variant thereof.
  • the binding polypeptide has the property of binding to netrin-1 and induce cell death or apoptosis of a tumor cell via an UNC5 receptor. The fact is that free or active netrin-1 does no longer exist or at an insufficient level, so that the apoptosis signaling of the netrin-1 receptor is activated.
  • the present invention also relates to a netrin-1 binding polypeptide which comprises one or more complementarity-determining region(s) (CDR(s)) having an amino acid sequence SEQ ID NO: 7, SEQ ID NO: 30 or SEQ ID NO: 9, wherein the binding polypeptide has the property of binding to netrin-1 and inducing cell death or apoptosis of a tumor cell via an UNC5 receptor.
  • CDR(s) complementarity-determining region(s)
  • the polypeptide may be an antibody or an epitope-binding fragment thereof.
  • the present invention thus relates to a netrin-1 binding polypeptide which comprises one or more complementarity-determining region (CDR) having an amino acid sequence SEQ ID NO: 7 (CDR3-H), SEQ ID NO: 9 (CDR3-L), and preferably both.
  • CDR complementarity-determining region
  • the polypeptide may be further defined by the additional presence of CDR1, CDR2 or the CDR1 and CDR2 of the VH and/or VL. Therefore, the polypeptide may comprise one or more CDR(s) having the amino acid sequences SEQ ID NO: 7 and SEQ ID NO: 5; SEQ ID NO: 7 and SEQ ID NO: 6; SEQ ID NO: 9 and the sequence YAS; and/or SEQ ID NO: 9 and SEQ ID NO: 8.
  • An object of the invention is a polypeptide comprising a CDR1-H of sequence SEQ ID NO: 5, a CDR2-H of sequence SEQ ID NO: 6, a CDR3-H of sequence SEQ ID NO: 7.
  • An object of the invention is a polypeptide comprising a CDR1-L of sequence SEQ ID NO: 8, a CDR2-L of sequence YAS and a CDR3-L of sequence SEQ ID NO: 9.
  • the polypeptide of the invention preferably comprises a CDR1-H of sequence SEQ ID NO: 5, a CDR2-H of sequence SEQ ID NO: 6, a CDR3-H of sequence SEQ ID NO: 7, a CDR1-L of sequence SEQ ID NO: 8, a CDR2-L of sequence YAS and a CDR3-L of sequence SEQ ID NO: 9.
  • the present invention thus relates to a netrin-1 binding polypeptide which comprises one or more complementarity-determining region (CDR) having an amino acid sequence SEQ ID NO: 30 (CDR3-H), SEQ ID NO: 9 (CDR3-L), and preferably both.
  • CDR complementarity-determining region
  • the polypeptide may be further defined by the additional presence of CDR1, CDR2 or the CDR1 and CDR2 of the VH and/or VL. Therefore, the polypeptide may comprise one or more CDR(s) having the amino acid sequences SEQ ID NO: 30 and SEQ ID NO: 28; SEQ ID NO: 30 and SEQ ID NO: 29; SEQ ID NO: 9 and the SEQ ID NO: 32; and/or SEQ ID NO: 9 and SEQ ID NO: 31.
  • An object of the invention is a polypeptide comprising a CDR1-H of sequence SEQ ID NO: 28, a CDR2-H of sequence SEQ ID NO: 29, a CDR3-H of sequence SEQ ID NO: 30.
  • An object of the invention is a polypeptide comprising a CDR1-L of sequence SEQ ID NO: 31, a CDR2-L of sequence SEQ ID NO: 32 and a CDR3-L of sequence SEQ ID NO: 9.
  • the polypeptide of the invention preferably comprises a CDR1-H of sequence SEQ ID NO: 28, a CDR2-H of sequence SEQ ID NO: 29, a CDR3-H of sequence SEQ ID NO: 30, a CDR1-L of sequence SEQ ID NO: 31, a CDR2-L of sequence SEQ ID NO:32 and a CDR3-L of sequence SEQ ID NO: 9.
  • polypeptides are netrin-1 binding polypeptides, wherein the binding polypeptide has the property of binding to netrin-1 and induce cell death or apoptosis of a tumor cell via an UNC5 receptor.
  • These polypeptides are preferably antibodies, especially monoclonal antibodies.
  • Various forms of binding polypeptides or antibodies (including fragments and combination thereof) will be described later herein.
  • the polypeptide or antibody of the invention comprises an amino acid sequence SEQ ID NO: 10, 11, 12 or 13. Typically, it comprises both sequences SEQ ID NO: 10 and 11, or SEQ ID NO: 12 and 13.
  • the polypeptide or antibody is humanized and comprises an amino acid sequence selected from the group of SEQ ID NO: 14 to 19 and/or from the group of SEQ ID NO: 20 to 27.
  • the polypeptide or antibody is humanized and comprises an amino acid sequence selected from the group of SEQ ID NO: 14 to 19 and an amino acid sequence selected from the group of SEQ ID NO: 20 to 27.
  • Another object of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising at least one netrin-1 binding polypeptide or antibody according to the invention and a pharmaceutically acceptable vehicle or excipient.
  • the polypeptide or antibody is humanized.
  • Still another object of the invention is a method of treatment of cancer wherein a subject in need thereof is administered with a therapeutically effective amount of a pharmaceutical composition comprising at least one netrin-1 binding polypeptide or antibody according to the invention and a pharmaceutically acceptable vehicle or excipient.
  • a pharmaceutical composition comprising at least one netrin-1 binding polypeptide or antibody according to the invention and a pharmaceutically acceptable vehicle or excipient.
  • the polypeptide or antibody is humanized.
  • composition and method may comprise any one of the features or combination of features as disclosed with respect to the polypeptide or antibody as disclosed herein.
  • the cancer is one wherein tumoral cells express or over-express a netrin-1 receptor, in particular of the UNC5 class, especially UNC5B and/or UNC5A.
  • a netrin-1 receptor in particular of the UNC5 class, especially UNC5B and/or UNC5A.
  • the tumoral cells escape netrin-1 receptor related apoptosis owing binding of netrin-1 to said receptor, in particular of the UNC5 class, especially UNC5B and/or UNC5A, in the presence of netrin-1.
  • the cancer is one wherein tumoral cells express or over-express netrin-1.
  • cancers include metastatic breast cancer, non-small cell lung cancer, aggressive neuroblastoma, pancreatic adenocarcinoma, primary melanoma, melanoma metastasis, ovarian cancers, glioblastoma, acute myeloid leukaemia, chronic lymphocytic leukaemia, aggressive B-cell lymphoma, sarcoma, renal adenocarcinoma, head and neck cancers, testicular cancers (e.g. embryonal carcinoma, teratoma, yolk sac tumors), kidney cancers, stomach cancers, uterus cancers
  • testicular cancers e.g. embryonal carcinoma, teratoma, yolk sac tumors
  • Methods of determining whether a given cell expresses netrin-1 dependence receptors DCC and/or UNC5 on the surface or shows significant up-regulation of netrin-1 gene expression are well known in the art and comprise, but are not limited to, IHC (Immunohistochemistry) of FACS (Fluorescence activated cell sorting), quantitative PCR (e.g.
  • compositions and methods are for the treatment of cancers expressing or over-expressing netrin-1, wherein this expression or over-expression is linked to the cancer itself, or is induced by a chemotherapeutic drug treatment alone, or both.
  • An object of the invention is a method of combined anti-cancer treatment comprising the administration to a patient in need thereof of a chemotherapeutic drug and of a polypeptide or antibody as disclosed herein.
  • the chemotherapeutic drug and the polypeptide or antibody are in effective amount.
  • Another object of the invention is a composition comprising a polypeptide or antibody as disclosed herein for use as an anti-cancer medicament to be used in combination with a chemotherapeutic drug in a patient.
  • the invention also relates to a composition comprising a polypeptide or antibody as disclosed herein for use as an anti-cancer medicament in a patient who is treated with a chemotherapeutic drug.
  • Another object of the invention is a composition comprising a chemotherapeutic drug for use as an anti-cancer medicament to be used in a patient in combination with a polypeptide or antibody as disclosed herein.
  • the invention also relates to a composition comprising a chemotherapeutic drug for use as an anti-cancer medicament in a patient who is treated with a polypeptide or antibody as disclosed herein.
  • Another object of the invention is a composition or kit of parts comprising a chemotherapeutic drug and a polypeptide or antibody as disclosed herein, for a simultaneous, separate or sequential administration to a patient.
  • Another object of the invention is a composition or kit of parts comprising a chemotherapeutic drug and a polypeptide or antibody as disclosed herein, for a simultaneous, separate or sequential administration to a patient, for use as an anti-cancer medicament or anti-cancer treatment.
  • Another object of the invention is a composition comprising a chemotherapeutic drug and a polypeptide or antibody as disclosed herein, in a pharmaceutically acceptable carrier or vehicle.
  • Another object of the invention is a composition comprising a chemotherapeutic drug and a polypeptide or antibody as disclosed herein, in a pharmaceutically acceptable carrier or vehicle, for use as an anti-cancer medicament.
  • Still another object is the use of a polypeptide or antibody as disclosed herein for the preparation of an anti-cancer medicament intended for a combined treatment of a patient with a chemotherapeutic drug.
  • Still another object is the use of a chemotherapeutic drug for the preparation of an anti-cancer medicament intended for a combined treatment of a patient with a polypeptide or antibody as disclosed herein.
  • Still another object is the use of a polypeptide or antibody as disclosed herein and a chemotherapeutic drug for the preparation of a combined anti-cancer medicament.
  • Still another object is the use of a polypeptide or antibody as disclosed herein and a chemotherapeutic drug for the preparation of a combined anti-cancer medicament composition or kit of parts, for a simultaneous, separate or sequential administration to a patient.
  • the chemotherapeutic drug is a drug which induces an over-expression of netrin-1 in cancer cells and the polypeptide or antibody as disclosed herein promotes netrin-1 receptors-induced apoptosis or cell death despite this overexpression.
  • the chemotherapeutic drug is in particular a drug which induces an over-expression of netrin-1 in cancer cells.
  • the determination that a drug induces a netrin-1 over-expression may be easily performed on any cancerous cell, such as cell line or cells from a biopsy.
  • the assay is performed on cells from the cancer to be treated, for example from a biopsy.
  • the assay is performed on a cell, such as a cell line, which is representative for the cancer to be treated.
  • the assay is made on a A549 or H460 cell line.
  • the assay may comprise comparing the netrin-1 gene expression between the cells treated with the chemotherapeutic drug and the cells not treated.
  • the expression may be measured by PCR, especially quantitative RT-PCR, for example using the primers disclosed and provided herein (SEQ ID NO: 33 and 34) as described in PCT/EP2013/068937 , the whole content of which is incorporated by reference or the skilled person may refer..
  • the classification of a drug in the family of those inducing this over-expression may simply be performed in accordance with the method described in the following Material and Method on a A549 or H460 cell line, as described in PCT/EP2013/068937 .
  • the chemotherapeutic drug is especially a cytotoxic drug.
  • the drug is doxorubicin, 5-fluorouracil (5FU), paclitaxel (e.g. Taxol), or cisplatin.
  • the drug is a cytotoxic antibiotic.
  • the cytotoxic antibiotic may be actinomycin, an anthracycline, bleomycin, plicamycin or mitomycin.
  • the anthracycline may be doxorubicin, daunorubicin, valrubicin, idarubicine or epirubicine.
  • the drug is an alkylating agent.
  • the alkylating agent may be a platinum derivative, such as cisplatin, carboplatin, oxaliplatine or other alkylating agents such as cyclophosphamide, ifosfamide, melphalan, thiotepa.
  • Other classes include-epipodophylotoxines, e.g. etoposide, topoisomerase inhibitors (camptotecines), e.g. irinotecan, topotecan , alkylating agents of the minor groove of DNA , e.g. Trabectedine (YONDELIS), methotrexate, pemetrexed, raltitrexed.
  • the drug is a taxane or other tubulin targeting agents.
  • the taxane may be paclitaxel or docetaxel, or eribuline (recently approved for breast cancer).
  • the drug is an antineoplastic agent such as :
  • a conservative amino acid substitution is one in which an amino acid residue is substituted by another amino acid residue having a side chain group with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein.
  • Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartic acid and glutamic acid; and 7) sulfur-containing side chains: cysteine and methionine.
  • Conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine-tryptophane, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.
  • An " antibody” may be a natural or conventional antibody in which two heavy chains are linked to each other by disulfide bonds and each heavy chain is linked to a light chain by a disulfide bond.
  • Each chain contains distinct sequence domains.
  • the light chain includes two domains or regions, a variable domain (VL) and a constant domain (CL).
  • the heavy chain includes four domains, a variable domain (VH) and three constant domains (CH1, CH2 and CH3, collectively referred to as CH).
  • variable regions of both light (VL) and heavy (VH) chains determine binding recognition and specificity to the antigen.
  • the constant region domains of the light (CL) and heavy (CH) chains confer important biological properties such as antibody chain association, secretion, trans-placental mobility, complement binding, and binding to Fc receptors (FcR).
  • the Fv fragment is the N-terminal part of the Fab fragment of an immunoglobulin and consists of the variable portions of one light chain and one heavy chain.
  • the specificity of the antibody resides in the structural complementarity between the antibody combining site and the antigenic determinant.
  • Antibody combining sites are made up of residues that are primarily from the hypervariable or complementarity determining regions (CDRs). Occasionally, residues from nonhypervariable or framework regions (FR) influence the overall domain structure and hence the combining site.
  • Complementarity Determining Regions or " CDRs” refer to amino acid sequences which together define the binding affinity and specificity of the natural Fv region of a native immunoglobulin binding site.
  • the light and heavy chains of an immunoglobulin each have three CDRs, designated CDR1-L, CDR2-L, CDR3-L and CDR1-H, CDR2-H, CDR3-H, respectively.
  • a conventional antibody antigen-binding site therefore, includes six CDRs, comprising the CDR set from each of a heavy and a light chain V region.
  • FRs Framework Regions
  • the light and heavy chains of an immunoglobulin each have four FRs, designated FR1-L, FR2-L, FR3-L, FR4-L, and FR1-H, FR2-H, FR3-H, FR4-H, respectively.
  • a "human framework region” is a framework region that is substantially identical (about 85%, or more, in particular 90%, 95%, 97%, 99% or 100%) to the framework region of a naturally occurring human antibody.
  • CDR/FR definition in an immunoglobulin light or heavy chain is to be determined based on IMGT definition (Lefranc et al. (2003) Dev Comp Immunol. 27 (1):55-77; www.imgt.org).
  • antibody denotes conventional antibodies and fragments thereof, as well as single domain antibodies and fragments thereof, in particular variable heavy chain of single domain antibodies, and chimeric, humanised, bispecific or multispecific antibodies.
  • antibody or immunoglobulin also includes " single domain antibodies " which have been more recently described and which are antibodies whose complementary determining regions are part of a single domain polypeptide.
  • single domain antibodies include heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional four-chain antibodies, engineered single domain antibodies.
  • Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, Ilama, goat, rabbit and bovine.
  • Single domain antibodies may be naturally occurring single domain antibodies known as heavy chain antibody devoid of light chains.
  • Camelidae species for example camel, dromedary, Ilama, alpaca and guanaco, produce heavy chain antibodies naturally devoid of light chain.
  • Camelid heavy chain antibodies also lack the CH1 domain.
  • VHH variable heavy chain of these single domain antibodies devoid of light chains
  • VHHs Similar to conventional VH domains, VHHs contain four FRs and three CDRs.
  • Nanobodies have advantages over conventional antibodies: they are about ten times smaller than IgG molecules, and as a consequence properly folded functional nanobodies can be produced by in vitro expression while achieving high yield. Furthermore, nanobodies are very stable, and resistant to the action of proteases. The properties and production of nanobodies have been reviewed by Harmsen and De Haard ( Harmsen and De Haard (2007) Appl. Microbiol. Biotechnol. 77:13-22 ).
  • monoclonal antibody or " mAb” as used herein refers to an antibody molecule of a single amino acid composition that is directed against a specific antigen, and is not to be construed as requiring production of the antibody by any particular method.
  • a monoclonal antibody may be produced by a single clone of B cells or hybridoma, but may also be recombinant, i.e. produced by protein engineering.
  • Fragments of (conventional) antibodies comprise a portion of an intact antibody, in particular the antigen binding region or variable region of the intact antibody.
  • antibody fragments include Fv, Fab, F(ab') 2 , Fab', dsFv, (dsFv) 2 , scFv, sc(Fv) 2 , diabodies, bispecific and multispecific antibodies formed from antibody fragments.
  • a fragment of a conventional antibody may also be a single domain antibody, such as a heavy chain antibody or VHH.
  • Fab denotes an antibody fragment having a molecular weight of about 50,000 Da and antigen binding activity, in which about a half of the N-terminal side of H chain and the entire L chain, among fragments obtained by treating IgG with a protease, papaine, are bound together through a disulfide bond.
  • F(ab') 2 refers to an antibody fragment having a molecular weight of about 100,000 Da and antigen binding activity, which is slightly larger than the Fab bound via a disulfide bond of the hinge region, among fragments obtained by treating IgG with a protease, pepsin.
  • a single chain Fv (“scFv ”) polypeptide is a covalently linked VH::VL heterodimer which is usually expressed from a gene fusion including VH and VL encoding genes linked by a peptide-encoding linker.
  • the human scFv fragment of the invention includes CDRs that are held in appropriate conformation, in particular by using gene recombination techniques.
  • Divalent and multivalent antibody fragments can form either spontaneously by association of monovalent scFvs, or can be generated by coupling monovalent scFvs by a peptide linker, such as divalent sc(Fv) 2 .
  • dsFv is a VH::VL heterodimer stabilised by a disulphide bond.
  • (dsFv) 2 denotes two dsFv coupled by a peptide linker.
  • bispecific antibody or "BsAb” denotes an antibody which combines the antigen-binding sites of two antibodies within a single molecule. Thus, BsAbs are able to bind two different antigens simultaneously. Genetic engineering has been used with increasing frequency to design, modify, and produce antibodies or antibody derivatives with a desired set of binding properties and effector functions as described for instance in EP 2 050 764 A1 .
  • multispecific antibody denotes an antibody which combines the antigen-binding sites of two or more antibodies within a single molecule.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL).
  • VH heavy-chain variable domain
  • VL light-chain variable domain
  • linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • the epitope-binding fragment is selected from the group consisting of Fv, Fab, F(ab') 2 , Fab', dsFv, (dsFv) 2 , scFv, sc(Fv) 2 , diabodies and VHH.
  • a “chimeric antibody”, as used herein, is an antibody in which the constant region, or a portion thereof, is altered, replaced, or exchanged, so that the variable region is linked to a constant region of a different species, or belonging to another antibody class or subclass.
  • Chimeric antibody also refers to an antibody in which the variable region, or a portion thereof, is altered, replaced, or exchanged, so that the constant region is linked to a variable region of a different species, or belonging to another antibody class or subclass.
  • humanised antibody refers to an antibody which is initially wholly or partially of non-human origin and which has been modified to replace certain amino acids, in particular in the framework regions of the heavy and light chains, in order to avoid or minimize an immune response in humans.
  • the constant domains of a humanized antibody are most of the time human CH and CL domains.
  • a humanized antibody has constant domains of human origin.
  • humanized antibody refers to a chimeric antibody which contain minimal sequence derived from non-human immunoglobulin, e.g. the CDRs.
  • polypeptide or "netrin-1 binding polypeptide” is used to encompass all these kinds of antibodies, fragments or combination thereof.
  • Humanized antibodies or antibodies adapted for non-rejection by other mammals, may be produced using several technologies such as resurfacing and CDR grafting.
  • the resurfacing technology uses a combination of molecular modeling, statistical analysis and mutagenesis to alter the non-CDR surfaces of antibody variable regions to resemble the surfaces of known antibodies of the target host.
  • Antibodies can be humanized using a variety of other techniques including CDR-grafting ( EP0239400 ; WO91/09967 ; U.S. Patent Nos. 5,530,101 and 5,585,089 ), veneering or resurfacing ( EP0592106 ; EP0519596 ; Padlan (1991) Molecular Immunology 28(4/5):489-498 ; Studnicka et al. (1994) Protein Engineering 7(6):805-814 ; Roguska et al. (1994) Proc. Natl. Acad. Sci U.S.A. 91:969-973 ), and chain shuffling ( U.S. Patent No. 5,565,332 ).
  • Human antibodies can be made by a variety of methods known in the art including phage display methods. See also U.S. Patent Nos. 4,444,887 , 4,716,111 , 5,545,806 , and 5,814,318 ; and International patent application WO98/46645 , WO98/50433 , WO98/24893 , WO98/16654 , WO96/34096 , WO96/33735 , and WO91/10741 .
  • treating means reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or one or more symptoms of such disorder or condition.
  • treating cancer as used herein is meant the inhibition of the growth of malignant cells of a tumour and/or the progression of metastases from said tumor.
  • Such treatment can also lead to the regression of tumor growth, i.e., the decrease in size of a measurable tumor.
  • such treatment leads to a partial regression of the tumor or metastasis.
  • such treatment leads to the complete regression of the tumor or metastasis.
  • the term "patient” or “patient in need thereof” is intended for a human or non-human mammal affected or likely to be affected with a malignant tumor.
  • the patient to be treated may have been previously treated with other anti-cancer treatments.
  • the patient to be treated may have been previously treated with an oxaliplatin-, cisplatin-, a carboplatin-, and/or a paclitaxel-docetaxel-based regimen.
  • a “therapeutically effective amount” of the polypeptide or antibody of the invention is meant a sufficient amount thereof to treat said cancer disease, at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the polypeptide or antibody of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific polypeptide or antibody employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific polypeptide or antibody employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide or antibody employed; and like factors well known in the medical arts.
  • said therapeutically effective amount of the polypeptide or antibody administered to the patient is a dose ranging from 5 mg/m 2 to 500 mg/m 2 , more particularly ranging from 150 mg/m 2 to 450 mg/m 2 of body surface area.
  • the polypeptide or antibody of the invention is administered repeatedly according to a protocol that depends on the patient to be treated (age, weight, treatment history, etc.), which can be determined by a skilled physician.
  • compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • compositions including the polypeptide or antibody of the invention and the route of administration naturally depend upon the condition to be treated, the severity of the illness, the age, weight, and gender of the patient, etc.
  • polypeptide or antibody of the invention can be formulated for a topical, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous or intraocular administration and the like.
  • the polypeptide or antibody of the invention is administered intravenously
  • the pharmaceutical compositions including the polypeptide or antibody of the invention may contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • an effective amount of the polypeptide or antibody of the invention may be dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like) and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants, stabilizing agents, cryoprotectants or antioxidants.
  • the prevention of the action of microorganisms can be brought about by antibacterial and antifungal agents. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 mL of isotonic NaCl solution and either added to 1000 mL of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, " Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580 ).
  • Example 1 ELISA-type binding assay of 4C11 antibodies to Netrin-1 ( Figure 1)
  • White 96-well microtiter plate (Costar 3912 Corning) was incubated overnight at 4°C with 100ng of His-tagged Netrin-1 (R&D 6419-N1) in 100 ⁇ L of Phosphate buffer saline (PBS). After three washings with 300 ⁇ L of PBS-0.05% Tween-20 (PBS-T), the plate was blocked by addition of 100 ⁇ L of PBS-3% BSA and incubated 1 hour at room temperature. After three washings with 300 ⁇ L of PBS-T, the plate was incubated with various quantities (10 ng to 1200ng) of the anti-netrin-1 antibody.
  • a relevant secondary antibody conjugated to horse radish peroxidase e.g. goat anti-human IgG (Fc) Sigma A0170 or goat anti-mouse IgG light chain specific (kappa) Jackson Immunoresearch 115-035-174
  • horse radish peroxidase e.g. goat anti-human IgG (Fc) Sigma A0170 or goat anti-mouse IgG light chain specific (kappa) Jackson Immunoresearch 115-035-174
  • HRP ECL western blotting substrate, PIERCE
  • Figure 1 shows the typical dose-dependent interaction of the 4C11 murine antibody to adsorbed netrin-1 in the ELISA-type assay.
  • the concentration of the antibodies allowing 50% binding are calculated from the sigmoidal binding curves as displayed in figure 1 .
  • Table 1 displays the EC 50 values (in ⁇ g/mL and in nM) of the different murine (4C11 whole antibody as well as Fab and Fab'2 fragments) and humanized 4C11 antibodies.
  • Table 1 Potency of the various 4C11 variants to bind to netrin-1 or to impair the netrin-1 binding onto UNC5B: Potency Data Binding (EC50) Ligand binding inhibition (IC50) ng/mL pM ng/mL pM 4C11 murin IgG1 57 380 75 502 4C11 murin IgG2a 63 420 85 567 4C11 Fab 650 13000 844 16880 4C11 Fab'2 35 350 89 890 HUM01 134 894 54 360 HUM03 166 1107 65 434 HUM09 153 1021 71 474 HUM08 134 894 77 514 HUM06 139 927 82 547 HUM05 151 1007 85 567 HUM07 125 834 89 594 HUM10 211 1407 110 734 HUM04 198 1321 199 1327 HUM02 260 1734 215 1434
  • Murine 4C11 (IgG1) antibody (SEQ ID NO: 12 and 13) was captured via amine coupled capture molecules. A series with increasing concentrations of netrin-1 was injected. Chip surface with amine coupled capture molecule alone was used as reference control surface for correction of possible buffer-effects or non-specific binding of netrin-I.
  • Capture molecules Anti-mouse IgG antibodies (from goat, Jackson Immuno Research).
  • Capture buffer PBS (pH7.4), 0.005% Tween 20
  • Running buffer 20mM Hepes pH7.4, 600mM NaCl, 0.005% Tween 20. The analyte was injected for 3 minutes followed by a dissociation phase of 90s.
  • SPR surface plasmon resonance
  • Example 3 4C11 inhibits the binding of Netrin-1 to UNC5B ( Figure 2)
  • White 96-well microtiter plate (Costar 3912 Corning) was incubated overnight at 4°C with either 100ng UNC5B-Fc (R&D 1006-UN-050) or DCC-Fc (R&D 844-DC-050) in 100 ⁇ L of Phosphate buffer saline (PBS). After three washings with 300 ⁇ L of PBS-0.05% Tween-20 (PBS-T), the plate was blocked by addition of 100 ⁇ L of PBS-2% BSA and incubated 1 hour at room temperature.
  • PBS Phosphate buffer saline
  • the plate was incubated for 1 hour at room temperature, with 100 ⁇ L of PBS-1% BSA containing 50ng/mL FLAG-tagged netrin-1 (Adipogen) and various quantities (0,2 ng to 3000ng) of the 4C11 antibody.
  • HRP horse radish peroxidase
  • Figure 2 shows the typical dose-dependent inhibition of netrin-1 binding to UNC5B by increasing amounts of the murine 4C11 murine antibody in an ELISA-type assay.
  • the concentration of the antibodies allowing 50% inhibition are calculated from the sigmoidal binding curves as displayed in figure 5 .
  • Table 1 gives the IC 50 values (in ⁇ g/mL and in nM) of the different murine (4C11 whole antibody as well as Fab and Fab'2 fragments) and humanized 4C11 antibodies.
  • Epitope mapping was performed using an array of 590 15-amino acid linear peptides covering the whole sequence of human Netrin-1 (without the signal peptide) with a peptide-peptide overlap of 14 amino-acids.
  • Linear peptides were synthesized using standard Fmoc-chemistry and deprotected using trifluoric acid.
  • the 455-well credit-card format olypropylene cards containing the covalently linked peptides were incubated 30min at 25°C in PBST (PBS-1% Tween 80) containing 5% SQ (SQ, Super-Q, 4% horse serum (v/v), 5% ovalbumin (w/v) in PBST.
  • the peptides were incubated with the 4C11 (1 ⁇ g/mL) PBST-0.1% SQ. After washing, the peptides were incubated with a 1/1000 dilution of anti-mouse antibody peroxidase conjugate (SouthernBiotech) for one hour at 25° C. After washing, the peroxidase substrate 2,2'-azino-di-3-ethylbenzthiazoline sulfonate (ABTS) and 2 microlitres of 3 percent H2O2 are added. After one hour, the color development are quantified with a charge coupled device (CCD) - camera and an image processing system.
  • CCD charge coupled device
  • Figure 3 shows that the murine 4C11 interacts specifically with peptides included in the SEQ ID NO: 3: VACNCNLHARRCRFNMELYKLSGRKSGGVCLNCRHNTAGRHCH.
  • Figure 4 is a cartoon displaying the location of the epitope recognized by the murine 4C11 antibody. This epitope is carried by the second EGF-like domain of Netrin-1.
  • Example 6 4C11-induced caspase-3 in human A549 lung adenocarcinoma epithelial cells ( Figure 5)
  • Figure 5 shows that the murine 4C11 antibody induces caspase 3 activity in human A549 lung adenocarcinoma epithelial cells.
  • Example 7 in vivo 4C11-induced tumor growth inhibition of A549 (Human lung adenocarcinoma epithelial cells) ( Figure 6) and GRANTA (human mantle cell lymphoma cells) cells xenografts ( Figure 7).
  • A549 Human lung adenocarcinoma epithelial cells
  • GRANTA human mantle cell lymphoma cells
  • mice with A549 xenografts were treated with 5mg/kg of 4C11 once a week, while mice with GRANTA xenografts received lower dose of 4C11 (2mg/kg) once or twice a week.
  • Figures 6 and 7 demonstrated significant suppression of tumor growth of human A549 lung adenocarcinoma epithelial cells (A) and human GRANTA human mantle cell lymphoma (B) xenografted in immunodeficient mice.
  • the 4C11 antibody shows an inhibition of A549 and GRANTA tumor growth.
  • Example 8 Synergy between humanized 4C11 (hum03) and doxorubicine in rat osteosarcoma ( Figure 8).
  • a radiation-induced rat osteosarcoma has been transformed into a transplantable model gratfed in paratibial position after denudation of the periosteum (ref. Allouche M.et al. , 1980, Int. J. Cancer 26, 777-782 ).
  • a chemotherapy agent Dox, Dorubicin
  • Rats grafted with the osteosarcoma received twice a week, intra-peritoneal injection of hum03 (4.4mg/kg) or the vehicle (ctr).
  • Figure 8 shows that an inhibition of osteosarcoma growth is achieved in animals treated simultaneously with 4C11 and doxorubicin.
  • Example 9 Effet of murine 4C11 in Inflammatory model of spontaneous colon cancer generated by inflammation. (mouse model of IBD (Inflammatory Bowel Disease)-associated colorectal cancer)
  • mice were sacrificed at the beginning of the tenth week and the colon was removed for histological analysis.
  • Table 3 clearly shows that the treatment of the mice with the 4C11 antibody prevents or slow-down the development of inflammatory-driven colon adenocarcinomas.
  • Table 3 Effect of the anti-Netrin mAb 4C11 on inflammatory-driven colon tumors in vivo.
  • Mouse model of IBD (Inflammatory Bowel Disease)-associated colorectal cancer was generated as already described (Proc Natl Acad Sci U S A. 2009 Oct 6;106(40):17146-51). Mice were first treated with azoxymethane (AOM) and Dextran sulfate sodium (DSS) to induce colorectal cancer.
  • AOM azoxymethane
  • DSS Dextran sulfate sodium
  • cells are lysed by sonication in modified RIPA buffer (50mM Tris-HCl, pH7.5, 150mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM EDTA, protease inhibitor cocktail and 5mM DTT) and incubated 1h at 4°C.
  • modified RIPA buffer 50mM Tris-HCl, pH7.5, 150mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM EDTA, protease inhibitor cocktail and 5mM DTT
  • Cellular debris are pelletted by centrifugation (10.000g 15' at 4°C) and protein extracts (200 ⁇ g per lane) are loaded onto 10% SDS-polyacrylamide gels and blotted onto PVDF sheets (Millipore Corporation, Billerica, MA, U.S.A.).
  • Filters are blocked with 10% non-fat dried milk and 5% BSA in PBS/0.1% Tween 20 (PBS-T) over-night and then incubated for 2h with rabbit polyclonal ⁇ -netrin-1 (dilution 1:500, clone H104, Santa Cruz Biotechnology, Santa Cruz, CA, USA) and mouse monoclonal ⁇ -actin (Santa Cruz Biotechnologies) antibodies. After three washes with PBS-T, filters are incubated with the appropriate HRP-conjugated secondary antibody (1:10000, Jackson ImmunoResearch, Suffolk, UK) for 1h. Detection is performed using West Dura Chemiluminescence System (Pierce, Rockford, IL, U.S.A.).
  • Example 11 Example of cancers over-expressing netrin-1 and expressing DCC and/or UNC5A and/or B and/or C and/or D to be candidate for the treatment with a UNC5-TRAP.
  • the percentage of netrin-1 overexpressing cases is given for each type of cancers for which expression of netrin-1 and its receptors have been quantified.
  • RNA is extracted using NucleoSpin ® RNA II Kit (Macherey Nagel, Düren, Germany) according to manufacturer's protocol. RT-PCR reactions are performed with iScript ® cDNA Synthesis Kit (Biorad). One ⁇ g total RNA is reverse-transcribed using the following program: 25°C for 5 min, 42°C for 30 min and 85°C for 5 min.
  • the target transcripts are amplified in LightCycler ® 2.0 apparatus (Roche Applied Science), using the LightCycler FastStart DNA Master SYBR Green I Kit (Roche Applied Science).
  • target genes are normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and phosphoglycerate kinase (PGK) genes, used as housekeeping genes.
  • GPDH glyceraldehyde 3-phosphate dehydrogenase
  • PGK phosphoglycerate kinase
  • cells are lysed by sonication in modified RIPA buffer (50mM Tris-HCl, pH7.5, 150mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM EDTA, protease inhibitor cocktail and 5mM DTT) and incubated 1h at 4°C.
  • modified RIPA buffer 50mM Tris-HCl, pH7.5, 150mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM EDTA, protease inhibitor cocktail and 5mM DTT
  • Cellular debris are pelletted by centrifugation (10.000g 15' at 4°C) and protein extracts (200 ⁇ g per lane) are loaded onto 10% SDS-polyacrylamide gels and blotted onto PVDF sheets (Millipore Corporation, Billerica, MA, U.S.A.).
  • Filters are blocked with 10% non-fat dried milk and 5% BSA in PBS/0.1% Tween 20 (PBS-T) over-night and then incubated for 2h with rabbit polyclonal ⁇ -netrin-1 (dilution 1:500, clone H104, Santa Cruz Biotechnology, Santa Cruz, CA, USA) and mouse monoclonal ⁇ -actin (Santa Cruz Biotechnologies) antibodies. After three washes with PBS-T, filters are incubated with the appropriate HRP-conjugated secondary antibody (1:10000, Jackson ImmunoResearch, Suffolk, UK) for 1h. Detection is performed using West Dura Chemiluminescence System (Pierce, Rockford, IL, U.S.A.).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Endocrinology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
EP14305034.2A 2014-01-10 2014-01-10 Anti-netrin-1 antibody Withdrawn EP2893939A1 (en)

Priority Applications (30)

Application Number Priority Date Filing Date Title
EP14305034.2A EP2893939A1 (en) 2014-01-10 2014-01-10 Anti-netrin-1 antibody
PCT/EP2015/050306 WO2015104360A1 (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody
HUE15700118A HUE048088T2 (hu) 2014-01-10 2015-01-09 Új, netrin-1 elleni ellenanyag
NZ721975A NZ721975A (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody
JP2016545904A JP6586095B2 (ja) 2014-01-10 2015-01-09 新規な抗ネトリン1抗体
LTEP15700118.1T LT3092003T (lt) 2014-01-10 2015-01-09 Naujas anti-netrino-1 antikūnas
PT157001181T PT3092003T (pt) 2014-01-10 2015-01-09 Novo anticorpo anti-netrina 1
TW104100790A TWI699211B (zh) 2014-01-10 2015-01-09 新穎抗軸突導向因子-1之抗體
DK15700118.1T DK3092003T3 (da) 2014-01-10 2015-01-09 Hidtil ukendt anti-netrin-1-antistof
PL15700118T PL3092003T3 (pl) 2014-01-10 2015-01-09 Nowe przeciwciało przeciw netrynie-1
US15/110,612 US10494427B2 (en) 2014-01-10 2015-01-09 Anti-netrin-1 antibody
SI201531074T SI3092003T1 (sl) 2014-01-10 2015-01-09 Novo protitelo proti netrinu-1
CN201580007753.8A CN105979966B (zh) 2014-01-10 2015-01-09 新的抗神经诱向因子-1抗体
KR1020167018645A KR102360967B1 (ko) 2014-01-10 2015-01-09 신규 항-네트린-1 항체
EA201691212A EA034676B1 (ru) 2014-01-10 2015-01-09 Новое антитело против нетрина-1
MX2016009029A MX2016009029A (es) 2014-01-10 2015-01-09 Anticuerpo anti-netrina-1 novedoso.
ARP150100062A AR099068A1 (es) 2014-01-10 2015-01-09 Anticuerpo anti-netrina-1
BR112016015811-3A BR112016015811B1 (pt) 2014-01-10 2015-01-09 Anticorpo de ligação da netrina-1, composição farmacêutica compreendendo o mesmo e uso do mesmo
ES15700118T ES2770620T3 (es) 2014-01-10 2015-01-09 Anticuerpo anti-netrina 1 novedoso
RS20200058A RS59818B1 (sr) 2014-01-10 2015-01-09 Novo anti-netrin-1 antitelo
EP15700118.1A EP3092003B1 (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody
CA2936308A CA2936308C (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody
AU2015205575A AU2015205575B2 (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody
SG11201605562VA SG11201605562VA (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody
IL246603A IL246603B (en) 2014-01-10 2016-07-04 A new antibody against netrin-1
PH12016501339A PH12016501339A1 (en) 2014-01-10 2016-07-05 Novel anti-netrin-1 antibody
ZA2016/04640A ZA201604640B (en) 2014-01-10 2016-07-07 Novel anti-netrin-1 antibody
CL2016001753A CL2016001753A1 (es) 2014-01-10 2016-07-08 Anticuerpo monoclonal humanizado que se une específicamente a netrina-1, composiciones farmacéuticas y usos del anticuerpo para el tratamiento del cáncer
US16/657,200 US11648309B2 (en) 2014-01-10 2019-10-18 Anti-netrin-1 antibody
HRP20192313TT HRP20192313T1 (hr) 2014-01-10 2019-12-23 Novo anti-netrin-1 protutijelo

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
EP14305034.2A EP2893939A1 (en) 2014-01-10 2014-01-10 Anti-netrin-1 antibody

Publications (1)

Publication Number Publication Date
EP2893939A1 true EP2893939A1 (en) 2015-07-15

Family

ID=49958401

Family Applications (2)

Application Number Title Priority Date Filing Date
EP14305034.2A Withdrawn EP2893939A1 (en) 2014-01-10 2014-01-10 Anti-netrin-1 antibody
EP15700118.1A Active EP3092003B1 (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP15700118.1A Active EP3092003B1 (en) 2014-01-10 2015-01-09 Novel anti-netrin-1 antibody

Country Status (28)

Country Link
US (2) US10494427B2 (pl)
EP (2) EP2893939A1 (pl)
JP (1) JP6586095B2 (pl)
KR (1) KR102360967B1 (pl)
CN (1) CN105979966B (pl)
AR (1) AR099068A1 (pl)
AU (1) AU2015205575B2 (pl)
BR (1) BR112016015811B1 (pl)
CA (1) CA2936308C (pl)
CL (1) CL2016001753A1 (pl)
DK (1) DK3092003T3 (pl)
EA (1) EA034676B1 (pl)
ES (1) ES2770620T3 (pl)
HR (1) HRP20192313T1 (pl)
HU (1) HUE048088T2 (pl)
IL (1) IL246603B (pl)
LT (1) LT3092003T (pl)
MX (1) MX2016009029A (pl)
NZ (1) NZ721975A (pl)
PH (1) PH12016501339A1 (pl)
PL (1) PL3092003T3 (pl)
PT (1) PT3092003T (pl)
RS (1) RS59818B1 (pl)
SG (1) SG11201605562VA (pl)
SI (1) SI3092003T1 (pl)
TW (1) TWI699211B (pl)
WO (1) WO2015104360A1 (pl)
ZA (1) ZA201604640B (pl)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11648309B2 (en) * 2014-01-10 2023-05-16 Netris Pharma Anti-netrin-1 antibody

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR106543A1 (es) 2015-11-02 2018-01-24 Netris Pharma Terapia de combinación de agente neutralizante de ntn1 con fármacos que inhiben el control epigenético
CN110291105B (zh) * 2017-01-05 2024-03-01 奈特里斯药物公司 导蛋白-1干扰药物和免疫检查点抑制剂药物的组合治疗
US20230040568A1 (en) * 2019-10-04 2023-02-09 Albert Einstein College Of Medicine Kir3dl3 is an inhibitory receptor of the immune system and uses thereof
KR20240041326A (ko) 2021-07-27 2024-03-29 네트리 파르마 네트린-1 검출, 동반 테스트 및 방사선 기반 요법
EP4245772A1 (en) 2022-03-18 2023-09-20 Netris Pharma Anti-netrin-1 antibody to treat liver inflammation
EP4249509A1 (en) 2022-03-22 2023-09-27 Netris Pharma Anti-netrin-1 antibody against arthritis-associated pain
WO2023186968A1 (en) 2022-03-29 2023-10-05 Netris Pharma Novel mcl-1 inhibitor and combination of mcl-1 and a bh3 mimetic, such as a bcl-2 inhibitor

Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1998016654A1 (en) 1996-10-11 1998-04-23 Japan Tobacco, Inc. Production of a multimeric protein by cell fusion method
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
WO1998046645A2 (en) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Method for the production of antihuman antigen receptors and uses thereof
WO1998050433A2 (en) 1997-05-05 1998-11-12 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
FR2878165A1 (fr) * 2004-11-22 2006-05-26 Centre Nat Rech Scient Nouvelles utilisations des netrines
WO2007099133A1 (en) 2006-02-28 2007-09-07 Centre National De La Recherche Scientifique (Cnrs) Screening for anti-cancer compounds using netrin-1 activity
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
WO2010079230A1 (en) * 2009-01-09 2010-07-15 Centre National De La Recherche Scientifique (Cnrs) Method for the selection of endothelial cells death inducers via netrin-1 and its applications
WO2012025618A1 (en) 2010-08-26 2012-03-01 F. Hoffmann-La Roche Ag Recombinant fc-fusion protein of the fifth fibronectin type iii domain of dcc
WO2014041078A1 (en) * 2012-09-12 2014-03-20 Netris Pharma Recombinant fc-fusion protein of the two immunoglobulin domains of unc5
WO2014041088A2 (en) * 2012-09-12 2014-03-20 Netris Pharma Combined treatment with netrin-1 interfering drug and chemotherapeutic drug

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2001293870A1 (en) 2000-10-16 2002-04-29 Bayer Aktiengesellschaft Regulation of human netrin binding membrane receptor unc5h-1
US20060025335A1 (en) 2004-01-30 2006-02-02 Kinane Thomas B Netrin compositions and methods of using the same
JP2008506703A (ja) * 2004-07-14 2008-03-06 ユニバーシティ オブ ユタ リサーチ ファウンデーション ネトリン関連化合物および用途
US7999072B2 (en) 2004-11-22 2011-08-16 Centre National De La Recherche Scientifique (Cnrs) Mutated netrin-4, fragments thereof and their use as medicines
US20060153840A1 (en) 2005-01-12 2006-07-13 Anne Eichmann Methods for preventing or treating a condition or a disease associated with angiogenesis
WO2009141440A1 (en) 2008-05-21 2009-11-26 Centre National De La Recherche Scientifique (Cnrs) Netrin-1 overexpression as a biological marker and a survival factor for aggressive neuroblastoma
JP5147062B2 (ja) 2008-07-17 2013-02-20 Necトーキン株式会社 巻線部品
AR074369A1 (es) 2008-11-20 2011-01-12 Genentech Inc Anticuerpos anti-unc 5b (receptor de netrina) y metodos de uso
US8052960B2 (en) * 2009-01-20 2011-11-08 The Penn State Research Foundation Netrin-1 as a biomarker of injury and disease
CN105555311B (zh) * 2013-07-23 2021-10-08 诺瓦利克有限责任公司 稳定的抗体组合物
EP2893939A1 (en) * 2014-01-10 2015-07-15 Netris Pharma Anti-netrin-1 antibody

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1991009967A1 (en) 1989-12-21 1991-07-11 Celltech Limited Humanised antibodies
WO1991010741A1 (en) 1990-01-12 1991-07-25 Cell Genesys, Inc. Generation of xenogeneic antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
US5565332A (en) 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
EP0592106A1 (en) 1992-09-09 1994-04-13 Immunogen Inc Resurfacing of rodent antibodies
WO1996033735A1 (en) 1995-04-27 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO1998016654A1 (en) 1996-10-11 1998-04-23 Japan Tobacco, Inc. Production of a multimeric protein by cell fusion method
WO1998024893A2 (en) 1996-12-03 1998-06-11 Abgenix, Inc. TRANSGENIC MAMMALS HAVING HUMAN IG LOCI INCLUDING PLURAL VH AND Vλ REGIONS AND ANTIBODIES PRODUCED THEREFROM
WO1998046645A2 (en) 1997-04-14 1998-10-22 Micromet Gesellschaft Für Biomedizinische Forschung Mbh Method for the production of antihuman antigen receptors and uses thereof
WO1998050433A2 (en) 1997-05-05 1998-11-12 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
FR2878165A1 (fr) * 2004-11-22 2006-05-26 Centre Nat Rech Scient Nouvelles utilisations des netrines
WO2007099133A1 (en) 2006-02-28 2007-09-07 Centre National De La Recherche Scientifique (Cnrs) Screening for anti-cancer compounds using netrin-1 activity
EP1989546A1 (en) 2006-02-28 2008-11-12 Centre National De La Recherche Scientifique (Cnrs) Screening for anti-cancer compounds using netrin-1 activity
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
WO2010079230A1 (en) * 2009-01-09 2010-07-15 Centre National De La Recherche Scientifique (Cnrs) Method for the selection of endothelial cells death inducers via netrin-1 and its applications
WO2012025618A1 (en) 2010-08-26 2012-03-01 F. Hoffmann-La Roche Ag Recombinant fc-fusion protein of the fifth fibronectin type iii domain of dcc
WO2014041078A1 (en) * 2012-09-12 2014-03-20 Netris Pharma Recombinant fc-fusion protein of the two immunoglobulin domains of unc5
WO2014041088A2 (en) * 2012-09-12 2014-03-20 Netris Pharma Combined treatment with netrin-1 interfering drug and chemotherapeutic drug

Non-Patent Citations (35)

* Cited by examiner, † Cited by third party
Title
ACKERMANN, NATURE, vol. 386, 1997, pages 838 - 842
ALLOUCHE M, INT. J. CANCER, vol. 26, 1980, pages 777 - 782
BERNET, GASTROENTEROLOGY, vol. 133, 2007, pages 180 - 1848
BRAISTED J E ET AL: "Netrin-1 promotes thalamic axon growth and is required for proper development of the thalamocortical projection", JOURNAL OF NEUROSCIENCE, SOCIETY FOR NEUROSCIENCE, US, vol. 20, no. 15, 1 August 2000 (2000-08-01), pages 5792 - 5801, XP002591612, ISSN: 0270-6474 *
BREDESEN, CELL DEATH DIFFER, vol. 12, 2005, pages 1031 - 1043
DELLOYE-BOURGEOIS ET AL., J. EXP. MED., 2009
DELLOYE-BOURGEOIS ET AL., JNCI, 2009
DELLOYE-BOURGEOIS ET AL.: "Interference with netrin-1 and tumor cell death in non-small cell lung cancer.", JNCI, vol. 101, no. 4, 18 February 2009 (2009-02-18), pages 237 - 247, XP002537859, ISSN: 1460-2105, DOI: 10.1093/JNCI/DJN491 *
DELLOYE-BOURGEOIS, J NATL CANCER INST, vol. 101, 2009, pages 237 - 247
DELLOYE-BOURGEOIS, JEM, vol. 206, 2009, pages 833 - 847
FITAMANT ET AL., PNAS, 2008
FITAMANT JULIEN ET AL: "Netrin-1 expression confers a selective advantage for tumor cell survival in metastatic breast cancer", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, US, vol. 105, no. 12, 25 March 2008 (2008-03-25), pages 4850 - 4855, XP002537856, ISSN: 0027-8424, DOI: 10.1073/PNAS.0709810105 *
FITAMANT, PROC NATL ACAD SCI, vol. 105, 2008, pages 4850 - 4855
GEISBRECHT, J .BIOL. CHEM., vol. 278, 2003, pages 32561 - 32568
GENNARO: "Remington's Pharmaceutical Sciences 15th Edition,", pages: 1035 - 1038
GENNARO: "Remington's Pharmaceutical Sciences" 15th Edition,", pages: 1570 - 1580
HARMSEN; DE HAARD, APPL. MICROBIOL. BIOTECHNOL., vol. 77, 2007, pages 13 - 22
HONG, CELL, vol. 97, 1999, pages 927 - 941
KAUFMANN ET AL., CELLULAR ONCOLOGY, 2009
KEINO-MASU, CELL, vol. 87, 1996, pages 175 - 185
KINZLER, PROC NATL ACAD SCI, vol. 100, 1996, pages 4173 - 4178
KRUGER, J. NEUR., vol. 24, 2004, pages 10826 - 10834
LEFRANC, DEV COMP IMMUNOL., vol. 27, no. 1, 2003, pages 55 - 77, Retrieved from the Internet <URL:www.imgt.org>
LINK ET AL., ANNALS OF CHIR. ONCO., 2007
MEHLEN, CELL MOL LIFE SCI, vol. 61, 2004, pages 1854 - 1866
MEHLEN, NATURE, vol. 395, 1998, pages 801 - 804
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
NEUFERT C ET AL., NAT PROTOC, vol. 2, 2007, pages 1998 - 2004
PADLAN, MOLECULAR IMMUNOLOGY, vol. 28, no. 4/5, 1991, pages 489 - 498
PANASTASIOU, ONCOTARGET, 2011
PARADISI ET AL., EMBO MOL MED, vol. 5, 2013, pages 1821 - 1834
PROC NATL ACAD SCI U S A., vol. 106, no. 40, 6 October 2009 (2009-10-06), pages 17146 - 51
ROGUSKA ET AL., PROC. NATL. ACAD. SCI U.S.A., vol. 91, 1994, pages 969 - 973
SHIN, GASTROENTEROLOGY, vol. 133, 2007, pages 1849 - 1857
STUDNICKA ET AL., PROTEIN ENGINEERING, vol. 7, no. 6, 1994, pages 805 - 814

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11648309B2 (en) * 2014-01-10 2023-05-16 Netris Pharma Anti-netrin-1 antibody

Also Published As

Publication number Publication date
US10494427B2 (en) 2019-12-03
EP3092003A1 (en) 2016-11-16
PH12016501339B1 (en) 2016-08-15
WO2015104360A1 (en) 2015-07-16
JP6586095B2 (ja) 2019-10-02
PH12016501339A1 (en) 2016-08-15
DK3092003T3 (da) 2020-01-20
IL246603A0 (en) 2016-08-31
KR102360967B1 (ko) 2022-02-08
US20200079842A1 (en) 2020-03-12
TWI699211B (zh) 2020-07-21
PL3092003T3 (pl) 2020-05-18
TW201605476A (zh) 2016-02-16
ES2770620T3 (es) 2020-07-02
SI3092003T1 (sl) 2020-03-31
JP2017505607A (ja) 2017-02-23
AU2015205575A1 (en) 2016-07-21
BR112016015811B1 (pt) 2023-09-26
AU2015205575B2 (en) 2017-09-21
SG11201605562VA (en) 2016-08-30
US20180072800A1 (en) 2018-03-15
CA2936308A1 (en) 2015-07-16
CN105979966A (zh) 2016-09-28
HUE048088T2 (hu) 2020-05-28
EP3092003B1 (en) 2019-11-06
NZ721975A (en) 2022-07-29
EA201691212A1 (ru) 2016-12-30
BR112016015811A2 (pt) 2018-03-27
LT3092003T (lt) 2020-01-27
EA034676B1 (ru) 2020-03-05
CA2936308C (en) 2023-02-28
MX2016009029A (es) 2016-09-09
HRP20192313T1 (hr) 2020-03-20
PT3092003T (pt) 2020-02-03
RS59818B1 (sr) 2020-02-28
CL2016001753A1 (es) 2017-05-19
KR20160106074A (ko) 2016-09-09
CN105979966B (zh) 2021-06-08
ZA201604640B (en) 2019-03-27
IL246603B (en) 2020-01-30
AR099068A1 (es) 2016-06-29
US11648309B2 (en) 2023-05-16

Similar Documents

Publication Publication Date Title
US11648309B2 (en) Anti-netrin-1 antibody
KR102084806B1 (ko) 인테그린 αvβ6에 대한 항체 및 암을 치료하기 위한 그의 용도
EP2848631B1 (en) Use of an anti-ang2 antibody
US20220106401A1 (en) ANTI-EpCAM ANTIBODIES, COMPOSITIONS COMPRISING ANTI-EpCAM ANTIBODIES AND METHODS OF MAKING AND USING ANTI-EpCAM ANTIBODIES
WO2018067819A1 (en) Compositions and methods for treatment of cancers
JP2023089245A (ja) M(h)dm2/4に対する抗体、ならびにがんの診断および処置におけるそれらの使用
US20220323599A1 (en) Anti-bcma antibody conjugate, compositions comprising the same, and methods of making and using the same
CA3048347A1 (en) Combined treatment with netrin-1 interfering drug and immune checkpoint inhibitors drugs
KR20180079232A (ko) L1cam 단백질에 특이적으로 결합하는 항체 및 피리미딘 유사체 및/또는 플라틴계 항암제를 포함하는 암의 예방 또는 치료용 약학적 조성물
WO2023165561A1 (en) Anti-cd39 antibodies and use thereof
WO2024110956A1 (en) Monoclonal antibodies targeting the human cd147/emmprin protein
CA3235789A1 (en) Cancer specific plectin-1 specific antibodies and methods of use thereof
US20180099054A1 (en) Treatment with anti-efna4 antibody-drug conjugates

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20140110

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20160116